Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 222(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37278764

ABSTRACT

Using our newly developed ultrafast camera described in the companion paper, we reduced the data acquisition periods required for photoactivation/photoconversion localization microscopy (PALM, using mEos3.2) and direct stochastic reconstruction microscopy (dSTORM, using HMSiR) by a factor of ≈30 compared with standard methods, for much greater view-fields, with localization precisions of 29 and 19 nm, respectively, thus opening up previously inaccessible spatiotemporal scales to cell biology research. Simultaneous two-color PALM-dSTORM and PALM-ultrafast (10 kHz) single fluorescent-molecule imaging-tracking has been realized. They revealed the dynamic nanoorganization of the focal adhesion (FA), leading to the compartmentalized archipelago FA model, consisting of FA-protein islands with broad diversities in size (13-100 nm; mean island diameter ≈30 nm), protein copy numbers, compositions, and stoichiometries, which dot the partitioned fluid membrane (74-nm compartments in the FA vs. 109-nm compartments outside the FA). Integrins are recruited to these islands by hop diffusion. The FA-protein islands form loose ≈320 nm clusters and function as units for recruiting FA proteins.


Subject(s)
Focal Adhesions , Molecular Dynamics Simulation , Diffusion , Focal Adhesions/metabolism , Integrins/metabolism , Single Molecule Imaging , Cell Biology
2.
Nat Commun ; 12(1): 751, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33531495

ABSTRACT

Optogenetic approaches for studying neuronal functions have proven their utility in the neurosciences. However, optogenetic tools capable of inducing synaptic plasticity at the level of single synapses have been lacking. Here, we engineered a photoactivatable (pa)CaMKII by fusing a light-sensitive domain, LOV2, to CaMKIIα. Blue light or two-photon excitation reversibly activated paCaMKII. Activation in single spines was sufficient to induce structural long-term potentiation (sLTP) in vitro and in vivo. paCaMKII activation was also sufficient for the recruitment of AMPA receptors and functional LTP in single spines. By combining paCaMKII with protein activity imaging by 2-photon FLIM-FRET, we demonstrate that paCaMKII activation in clustered spines induces robust sLTP via a mechanism that involves the actin-regulatory small GTPase, Cdc42. This optogenetic tool for dissecting the function of CaMKII activation (i.e., the sufficiency of CaMKII rather than necessity) and for manipulating synaptic plasticity will find many applications in neuroscience and other fields.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Long-Term Potentiation/physiology , Optogenetics/methods , Synapses/metabolism , Animals , Electrophysiology , Female , HeLa Cells , Hippocampus/metabolism , Hippocampus/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Synapses/physiology
3.
PLoS One ; 12(11): e0188778, 2017.
Article in English | MEDLINE | ID: mdl-29190677

ABSTRACT

Electron tomography of the plasma membrane (PM) identified several layers of cortical actin meshwork running parallel to the PM cytoplasmic surface throughout the PM. Here, cortical actin structures and dynamics were examined in living cells, using super-resolution microscopy, with (x,y)- and z-resolutions of ~140 and ~400 nm, respectively, and single-molecule imaging. The super-resolution microscopy identified sub-micron-sized actin clusters that appeared identical by both phalloidin post-fixation staining and Lifeact-mGFP expression followed by fixation, and therefore, these actin clusters were named "actin-pl-clusters". In live cells, the actin-pl-clusters visualized by Lifeact-mGFP linked two or more actin filaments in the fine actin meshwork, acting as a node of the meshwork, and dynamically moved on/along the meshwork in a myosin II-dependent manner. Their formation depended on the Arp2/3 activities, suggesting that the movements could involve both the myosin motor activity and actin polymerization-depolymerization. The actin-pl-clusters differ from the actin nodes/asters found previously after latrunculin treatments, since myosin II and filamin A were not colocalized with the actin-pl-clusters, and the actin-pl-clusters were much smaller than the previously reported nodes/asters. The Lifeact linked to a fluorescently-labeled transmembrane peptide from syntaxin4 (Lifeact-TM) expressed in the PM exhibited temporary immobilization in the PM regions on which actin-pl-clusters and stress fibers were projected, showing that ≥66% of actin-pl-clusters and 89% of stress fibers were located in close proximity (within 3.5 nm) to the PM cytoplasmic surface. Podosome-associated cytoplasmic proteins, Tks4, Tks5, cortactin, and N-WASP, were transiently recruited to actin-pl-clusters, and thus, we propose that actin-pl-clusters also represent "actin podosome-like clusters".


Subject(s)
Actins/metabolism , Podosomes/metabolism , Single Molecule Imaging/methods , Animals , Cells, Cultured
4.
Sci Rep ; 7(1): 6791, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28754922

ABSTRACT

Fluorescence lifetime imaging microscopy (FLIM)-based Förster resonance energy transfer (FRET) measurement (FLIM-FRET) is one of the powerful methods for imaging of intracellular protein activities such as protein-protein interactions and conformational changes. Here, using saturation mutagenesis, we developed a dark yellow fluorescent protein named ShadowY that can serve as an acceptor for FLIM-FRET. ShadowY is spectrally similar to the previously reported dark YFP but has a much smaller quantum yield, greater extinction coefficient, and superior folding property. When ShadowY was paired with mEGFP or a Clover mutant (CloverT153M/F223R) and applied to a single-molecule FRET sensor to monitor a light-dependent conformational change of the light-oxygen-voltage domain 2 (LOV2) in HeLa cells, we observed a large FRET signal change with low cell-to-cell variability, allowing for precise measurement of individual cell responses. In addition, an application of ShadowY to a separate-type Ras FRET sensor revealed an EGF-dependent large FRET signal increase. Thus, ShadowY in combination with mEGFP or CloverT153M/F223R is a promising FLIM-FRET acceptor.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Mutation , Protein Domains
5.
Sci Rep ; 7: 46840, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28589959

ABSTRACT

This corrects the article DOI: 10.1038/srep15334.

6.
Cell Biochem Biophys ; 75(3-4): 399-412, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28646414

ABSTRACT

The central mechanism for the transmission of the prion protein misfolding is the structural conversion of the normal cellular prion protein to the pathogenic misfolded prion protein, by the interaction with misfolded prion protein. This process might be enhanced due to the homo-dimerization/oligomerization of normal prion protein. However, the behaviors of normal prion protein in the plasma membrane have remained largely unknown. Here, using single fluorescent-molecule imaging, we found that both prion protein and Thy1, a control glycosylphosphatidylinositol-anchored protein, exhibited very similar intermittent transient immobilizations lasting for a few seconds within an area of 24.2 and 3.5 nm in diameter in CHO-K1 and hippocampal neurons cultured for 1- and 2-weeks, respectively. Prion protein molecules were immobile during 72% of the time, approximately 1.4× more than Thy1, due to prion protein's higher immobilization frequency. When mobile, prion protein diffused 1.7× slower than Thy1. Prion protein's slower diffusion might be caused by its transient interaction with other prion protein molecules, whereas its brief immobilization might be due to temporary association with prion protein clusters. Prion protein molecules might be newly recruited to prion protein clusters all the time, and simultaneously, prion protein molecules in the cluster might be departing continuously. Such dynamic interactions of normal prion protein molecules would strongly enhance the spreading of misfolded prion protein.


Subject(s)
Cell Membrane/metabolism , Glycosylphosphatidylinositols/chemistry , Prion Proteins/metabolism , Thy-1 Antigens/metabolism , Animals , CHO Cells , Cell Membrane/chemistry , Cells, Cultured , Cricetinae , Cricetulus , Diffusion , Fluorescent Dyes/chemistry , Glycosylphosphatidylinositols/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Microscopy, Fluorescence , Prion Proteins/chemistry , Rats , Rats, Wistar , Thy-1 Antigens/chemistry
8.
Neuron ; 94(1): 37-47.e5, 2017 Apr 05.
Article in English | MEDLINE | ID: mdl-28318784

ABSTRACT

Elucidating temporal windows of signaling activity required for synaptic and behavioral plasticity is crucial for understanding molecular mechanisms underlying these phenomena. Here, we developed photoactivatable autocamtide inhibitory peptide 2 (paAIP2), a genetically encoded, light-inducible inhibitor of CaMKII activity. The photoactivation of paAIP2 in neurons for 1-2 min during the induction of LTP and structural LTP (sLTP) of dendritic spines inhibited these forms of plasticity in hippocampal slices of rodents. However, photoactivation ∼1 min after the induction did not affect them, suggesting that the initial 1 min of CaMKII activation is sufficient for inducing LTP and sLTP. Furthermore, the photoactivation of paAIP2 expressed in amygdalar neurons of mice during an inhibitory avoidance task revealed that CaMKII activity during, but not after, training is required for the memory formation. Thus, we demonstrated that paAIP2 is useful to elucidate the temporal window of CaMKII activation required for synaptic plasticity and learning.


Subject(s)
Avoidance Learning/physiology , CA1 Region, Hippocampal/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/metabolism , Neuronal Plasticity/physiology , Pyramidal Cells/metabolism , Animals , Animals, Newborn , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Dendritic Spines/physiology , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , HeLa Cells , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , Humans , Immunoblotting , Immunohistochemistry , Kinetics , Long-Term Potentiation/physiology , Mice , Microscopy, Fluorescence , Neurons/metabolism , Neurons/physiology , Optogenetics , Pyramidal Cells/physiology , RNA-Binding Proteins , Rats , Recombinant Fusion Proteins/genetics , Repressor Proteins , Tumor Suppressor Proteins/genetics
9.
Sci Rep ; 5: 15334, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26469148

ABSTRACT

Measurement of Förster resonance energy transfer by fluorescence lifetime imaging microscopy (FLIM-FRET) is a powerful method for visualization of intracellular signaling activities such as protein-protein interactions and conformational changes of proteins. Here, we developed a dark green fluorescent protein (ShadowG) that can serve as an acceptor for FLIM-FRET. ShadowG is spectrally similar to monomeric enhanced green fluorescent protein (mEGFP) and has a 120-fold smaller quantum yield. When FRET from mEGFP to ShadowG was measured using an mEGFP-ShadowG tandem construct with 2-photon FLIM-FRET, we observed a strong FRET signal with low cell-to-cell variability. Furthermore, ShadowG was applied to a single-molecule FRET sensor to monitor a conformational change of CaMKII and of the light oxygen voltage (LOV) domain in HeLa cells. These sensors showed reduced cell-to-cell variability of both the basal fluorescence lifetime and response signal. In contrast to mCherry- or dark-YFP-based sensors, our sensor allowed for precise measurement of individual cell responses. When ShadowG was applied to a separate-type Ras FRET sensor, it showed a greater response signal than did the mCherry-based sensor. Furthermore, Ras activation and translocation of its effector ERK2 into the nucleus could be observed simultaneously. Thus, ShadowG is a promising FLIM-FRET acceptor.


Subject(s)
Fluorescence Resonance Energy Transfer , Luminescent Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Microscopy, Fluorescence , Mutagenesis , Plasmids/metabolism , Protein Interaction Domains and Motifs , Quantum Theory , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , ras Proteins/chemistry , ras Proteins/genetics , ras Proteins/metabolism , Red Fluorescent Protein
10.
PLoS One ; 10(3): e0121109, 2015.
Article in English | MEDLINE | ID: mdl-25799407

ABSTRACT

Genetically encoded fluorescence resonance energy transfer (FRET) biosensors have been successfully used to visualize protein activity in living cells. The sensitivity and accuracy of FRET measurements directly depend on biosensor folding efficiency, expression pattern, sensitivity, and dynamic range. Here, to improve the folding efficiency of the Ca2+/calmodulin-dependent protein kinase II alpha (CaMKIIα) FRET biosensor, we amplified the association domain of the CaMKIIα gene using error-prone polymerase chain reaction (PCR) and fused it to the N-terminus of mCherry in a bacterial expression vector. We also created an Escherichia coli expression library based on a previously reported fluorescent protein folding reporter method, and found a bright red fluorescent colony that contained the association domain with four mutations (F394L, I419V, A430T, and I434T). In vitro assays using the purified mutant protein confirmed improved folding kinetics of the downstream fluorescent protein, but not of the association domain itself. Furthermore, we introduced these mutations into the previously reported CaMKIIα FRET sensor and monitored its Ca2+/calmodulin-dependent activation in HeLa cells using 2-photon fluorescence lifetime imaging microscopy (2pFLIM), and found that the expression pattern and signal reproducibility of the mutant sensor were greatly improved without affecting the autophosphorylation function and incorporation into oligomeric CaMKIIα. We believe that our improved CaMKIIα FRET sensor would be useful in various types of cells and tissues, providing data with high accuracy and reproducibility. In addition, the method described here may also be applicable for improving the performance of all currently available FRET sensors.


Subject(s)
Biosensing Techniques/methods , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Fluorescence Resonance Energy Transfer/methods , Mutation , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Models, Molecular , Protein Folding , Protein Structure, Tertiary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Red Fluorescent Protein
11.
Cytoskeleton (Hoboken) ; 70(3): 161-77, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23341328

ABSTRACT

The focal adhesion (FA) is an integrin-based structure built in/on the plasma membrane (PM), linking the extracellular matrix to the actin stress-fibers, working as cell migration scaffolds. Previously, we proposed the archipelago architecture of the FA, in which FA largely consists of fluid membrane, dotted with small islands accumulating FA proteins: membrane molecules enter the inter-island channels in the FA zone rather freely, and the integrins in the FA-protein islands rapidly exchanges with those in the bulk membrane. Here, we examined how Rac1, a small G-protein regulating FA formation, and its activators αPIX and ßPIX, are recruited to the FA zones. PIX molecules are recruited from the cytoplasm to the FA zones directly. In contrast, majorities of Rac1 molecules first arrive from the cytoplasm on the general inner PM surface, and then enter the FA zones via lateral diffusion on the PM, which is possible due to rapid Rac1 diffusion even within the FA zones, slowed only by a factor of two to four compared with that outside. The constitutively-active Rac1 mutant exhibited temporary and all-time immobilizations in the FA zone, suggesting that upon PIX-induced Rac1 activation at the FA-protein islands, Rac1 tends to be immobilized at the FA-protein islands.


Subject(s)
Cell Membrane/metabolism , Focal Adhesions/metabolism , Guanine Nucleotide Exchange Factors/metabolism , rac1 GTP-Binding Protein/metabolism , Cytoplasm/metabolism , HeLa Cells , Humans , Rho Guanine Nucleotide Exchange Factors
12.
Cytoskeleton (Hoboken) ; 69(6): 380-92, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22488960

ABSTRACT

The focal adhesion (FA) is an integrin-based structure built in/on the plasma membrane, mechanically linking the extracellular matrix with the termini of actin stress fibers, providing key scaffolds for the cells to migrate in tissues. The FA was considered as a micron-scale, massive assembly of various proteins, although its formation and decomposition occur quickly in several to several 10 s of minutes. The mechanism of rapid FA regulation has been a major mystery in cell biology. Here, using fast single fluorescent-molecule imaging, we found that transferrin receptor and Thy1, non-FA membrane proteins, readily enter the FA zone, diffuse rapidly there, and exit into the bulk plasma membrane. Integrin ß3 also readily enters the FA zone, and repeatedly undergoes temporary immobilization and diffusion in the FA zone, whereas approximately one-third of integrin ß3 is immobilized there. These results are consistent with the archipelago architecture of the FA, which consists of many integrin islands: the membrane molecules enter the inter-island channels rather freely, and the integrins in the integrin islands can be rapidly exchanged with those in the bulk membrane. Such an archipelago architecture would allow rapid FA formation and disintegration, and might be applicable to other large protein domains in the plasma membrane.


Subject(s)
Cell Membrane/metabolism , Focal Adhesions/metabolism , Proteins/metabolism , Animals , Biological Transport , Diffusion , HeLa Cells , Humans , Integrin beta3/metabolism , Models, Biological , Rats , Receptors, Transferrin/metabolism , Thy-1 Antigens/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...